1
|
Ridha Z, Fabi SG, Zubar R, Dayan SH. Decoding the Implications of Glucagon-like Peptide-1 Receptor Agonists on Accelerated Facial and Skin Aging. Aesthet Surg J 2024; 44:NP809-NP818. [PMID: 38874170 DOI: 10.1093/asj/sjae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/15/2024] Open
Abstract
Following the advent of glucagon-like peptide-1 receptor agonists (GLP-1RAs), subsequent unintended effects such as accelerated facial aging and altered skin health have been noted. This review delves deeper into the causative underlying mechanisms and provides insights into the intricate relationship between GLP-1RAs, adipose tissue, and premature facial aging, thereby highlighting the need for a nuanced understanding of their effects on facial alterations and skin health. Studies exploring the potential effects of GLP-1RAs on facial alterations and offering insights into the possible underlying mechanisms, causes, and clinical implications were included. The accelerated facial aging and altered skin health observed in GLP-1RA patients appears to be multifactorial, involving loss of dermal and subcutaneous white adipose tissue, and altered proliferation and differentiation of adipose-derived stem cells (ADSCs), and impacts on the production and secretion of hormonal and metabolic factors. These changes compromise the structural integrity and barrier function of the skin and may lead to diminished facial muscle mass, further exacerbating the appearance of aging. The insights presented call for a paradigm shift in the clinical management of facial changes induced by GLP-1RAs, with a focus on treatment strategies aimed at targeting ADSC stimulation. These include autologous fat transfers to reintroduce cells rich in ADSCs for rejuvenation, composite fat grafting combining autologous fat with/without stromal vascular fraction, and the strategic use of soft tissue fillers for volume restoration and biostimulation. This review highlights the potential role of GLP-1RAs in modulating adipose tissue dynamics, thereby contributing to accelerated aging through metabolic, structural, and hormonal pathways. LEVEL OF EVIDENCE: 5
Collapse
|
2
|
Alizadeh S, Mahboobi L, Nasiri M, Khosrowpour Z, Khosravimelal S, Asgari F, Gholipour-Malekabadi M, Taghi Razavi-Toosi SM, Singh Chauhan NP, Ghobadi F, Nasiri H, Gholipourmalekabadi M. Decellularized Placental Sponge Seeded with Human Mesenchymal Stem Cells Improves Deep Skin Wound Healing in the Animal Model. ACS APPLIED BIO MATERIALS 2024; 7:2140-2152. [PMID: 38470456 DOI: 10.1021/acsabm.3c00747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Skin injuries lead to a large burden of morbidity. Although numerous clinical and scientific strategies have been investigated to repair injured skin, optimal regeneration therapy still poses a considerable obstacle. To address this challenge, decellularized extracellular matrix-based scaffolds recellularized with stem cells offer significant advancements in skin regeneration and wound healing. Herein, a decellularized human placental sponge (DPS) was fabricated using the decellularization and freeze-drying technique and then recellularized with human adipose-derived mesenchymal cells (MSCs). The biological and biomechanical properties and skin full-thickness wound healing capacity of the stem cells-DPS constructs were investigated in vitro and in vivo. The DPS exhibited a uniform 3D microstructure with an interconnected pore network, 89.21% porosity, a low degradation rate, and good mechanical properties. The DPS and MSCs-DPS constructs were implanted in skin full-thickness wound models in mice. An accelerated wound healing was observed in the wounds implanted with the MSCs-DPS construct when compared to DPS and control (wounds with no treatment) during 7 and 21 days postimplantation follow-up. In the MSCs-DPS group, the wound was completely re-epithelialized, the epidermis layer was properly organized, and the dermis and epidermis' bilayer structures were restored after 7 days. Our findings suggest that DPS is an excellent carrier for MSC culture and delivery to skin wounds and now promises to proceed with clinical evaluations.
Collapse
Affiliation(s)
- Sanaz Alizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Leila Mahboobi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Modara Nasiri
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran 19585, Iran
- Research Company Located in Islamic Azad University Science and Technology Park, Araz Fidar Azma, Tehran, 1477893855, Iran
| | - Zahra Khosrowpour
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Sadjad Khosravimelal
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Fatemeh Asgari
- Avicenna Infertility Clinic, Avicenna Research Institute, ACECR, Tehran 1985743413, Iran
| | | | - Seyyed Mohammad Taghi Razavi-Toosi
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht 41887-94755, Iran
- Medical Biotechnology Research Center, Guilan University of Medical Sciences, Rasht 41887-94755, Iran
| | - Narendra Pal Singh Chauhan
- Department of Chemistry, Faculty of Science, Bhupal Nobles' University, Udaipur, Rajasthan 313001, India
| | - Faezeh Ghobadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran 14588-89694, Iran
| | - Hajar Nasiri
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| |
Collapse
|
3
|
Strauß S, Diemer M, Bucan V, Kuhbier JW, Asendorf T, Vogt PM, Schlottmann F. Spider silk enhanced tissue engineering of cartilage tissue: Approach of a novel bioreactor model using adipose derived stromal cells. J Appl Biomater Funct Mater 2024; 22:22808000241226656. [PMID: 38253568 DOI: 10.1177/22808000241226656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024] Open
Abstract
Human cartilage tissue remains a challenge for the development of therapeutic options due to its poor vascularization and reduced regenerative capacities. There are a variety of research approaches dealing with cartilage tissue engineering. In addition to different biomaterials, numerous cell populations have been investigated in bioreactor-supported experimental setups to improve cartilage tissue engineering. The concept of the present study was to investigate spider silk cocoons as scaffold seeded with adipose-derived stromal cells (ASC) in a custom-made bioreactor model using cyclic axial compression to engineer cartilage-like tissue. For chemical induction of differentiation, BMP-7 and TGF-β2 were added and changes in cell morphology and de-novo tissue formation were investigated using histological staining to verify chondrogenic differentiation. By seeding spider silk cocoons with ASC, a high colonization density and cell proliferation could be achieved. Mechanical induction of differentiation using a newly established bioreactor model led to a more roundish cell phenotype and new extracellular matrix formation, indicating a chondrogenic differentiation. The addition of BMP-7 and TGF-β2 enhanced the expression of cartilage specific markers in immunohistochemical staining. Overall, the present study can be seen as pilot study and valuable complementation to the published literature.
Collapse
Affiliation(s)
- Sarah Strauß
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Maximilian Diemer
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Jörn W Kuhbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
- Department of Plastic, Aesthetic and Hand Surgery, Helios Klinikum Hildesheim, Hildesheim, Germany
| | - Tomke Asendorf
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Frederik Schlottmann
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Luo L, Zhang W, Wang J, Zhao M, Shen K, Jia Y, Li Y, Zhang J, Cai W, Xiao D, Bai X, Liu K, Wang K, Zhang Y, Zhu H, Zhou Q, Hu D. A Novel 3D Culture Model of Human ASCs Reduces Cell Death in Spheroid Cores and Maintains Inner Cell Proliferation Compared With a Nonadherent 3D Culture. Front Cell Dev Biol 2021; 9:737275. [PMID: 34858974 PMCID: PMC8632442 DOI: 10.3389/fcell.2021.737275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
3D cell culture technologies have recently shown very valuable promise for applications in regenerative medicine, but the most common 3D culture methods for mesenchymal stem cells still have limitations for clinical application, mainly due to the slowdown of inner cell proliferation and increase in cell death rate. We previously developed a new 3D culture of adipose-derived mesenchymal stem cells (ASCs) based on its self-feeder layer, which solves the two issues of ASC 3D cell culture on ultra-low attachment (ULA) surface. In this study, we compared the 3D spheroids formed on the self-feeder layer (SLF-3D ASCs) with the spheroids formed by using ULA plates (ULA-3D ASCs). We discovered that the cells of SLF-3D spheroids still have a greater proliferation ability than ULA-3D ASCs, and the volume of these spheroids increases rather than shrinks, with more viable cells in 3D spheroids compared with the ULA-3D ASCs. Furthermore, it was discovered that the SLF-3D ASCs are likely to exhibit the abovementioned unique properties due to change in the expression level of ECM-related genes, like COL3A1, MMP3, HAS1, and FN1. These results indicate that the SLF-3D spheroid is a promising way forward for clinical application.
Collapse
Affiliation(s)
- Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Wei Zhang
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jing Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Ming Zhao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Jian Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Dan Xiao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Kaituo Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Yue Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Huayu Zhu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Qin Zhou
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, China
| |
Collapse
|
5
|
Bio-engineering a prevascularized human tri-layered skin substitute containing a hypodermis. Acta Biomater 2021; 134:215-227. [PMID: 34303011 DOI: 10.1016/j.actbio.2021.07.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Severe injuries to skin including hypodermis require full-thickness skin replacement. Here, we bioengineered a tri-layered human skin substitute (TLSS) containing the epidermis, dermis, and hypodermis. The hypodermal layer was generated by differentiation of human adipose stem cells (ASC) in a collagen type I hydrogel and combined with a prevascularized dermis consisting of human dermal microvascular endothelial cells and fibroblasts, which arranged into a dense vascular network. Subsequently, keratinocytes were seeded on top to generate the epidermal layer of the TLSS. The differentiation of ASC into adipocytes was confirmed in vitro on the mRNA level by the presence of adiponectin, as well as by the expression of perilipin and FABP-4 proteins. Moreover, functional characteristics of the hypodermis in vitro and in vivo were evaluated by Oil Red O, BODIPY, and AdipoRed stainings visualizing intracellular lipid droplets. Further, we demonstrated that both undifferentiated ASC and mature adipocytes present in the hypodermis influenced the keratinocyte maturation and homeostasis in the skin substitutes after transplantation. In particular, an enhanced secretion of TGF-β1 by these cells affected the epidermal morphogenesis as assessed by the expression of key proteins involved in the epidermal differentiation including cytokeratin 1, 10, 19 and cornified envelope formation such as involucrin. Here, we propose a novel functional hypodermal-dermo-epidermal tri-layered skin substitute containing blood capillaries that efficiently promote regeneration of skin defects. STATEMENT OF SIGNIFICANCE: The main objective of this study was to develop and assess the usefulness of a tri-layered human prevascularized skin substitute (TLSS) containing an epidermis, dermis, and hypodermis. The bioengineered hypodermis was generated from human adipose mesenchymal stem cells (ASC) and combined with a prevascularized dermis and epidermis. The TLSS represents an exceptional model for studying the role of cell-cell and cell-matrix interactions in vitro and in vivo. In particular, we observed that enhanced secretion of TGF-β1 in the hypodermis exerted a profound impact on fibroblast and keratinocyte differentiation, as well as epidermal barrier formation and homeostasis. Therefore, improved understanding of the cell-cell interactions in such a physiological skin model is essential to gain insights into different aspects of wound healing.
Collapse
|
6
|
Horder H, Guaza Lasheras M, Grummel N, Nadernezhad A, Herbig J, Ergün S, Teßmar J, Groll J, Fabry B, Bauer-Kreisel P, Blunk T. Bioprinting and Differentiation of Adipose-Derived Stromal Cell Spheroids for a 3D Breast Cancer-Adipose Tissue Model. Cells 2021; 10:cells10040803. [PMID: 33916870 PMCID: PMC8066030 DOI: 10.3390/cells10040803] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Biofabrication, including printing technologies, has emerged as a powerful approach to the design of disease models, such as in cancer research. In breast cancer, adipose tissue has been acknowledged as an important part of the tumor microenvironment favoring tumor progression. Therefore, in this study, a 3D-printed breast cancer model for facilitating investigations into cancer cell-adipocyte interaction was developed. First, we focused on the printability of human adipose-derived stromal cell (ASC) spheroids in an extrusion-based bioprinting setup and the adipogenic differentiation within printed spheroids into adipose microtissues. The printing process was optimized in terms of spheroid viability and homogeneous spheroid distribution in a hyaluronic acid-based bioink. Adipogenic differentiation after printing was demonstrated by lipid accumulation, expression of adipogenic marker genes, and an adipogenic ECM profile. Subsequently, a breast cancer cell (MDA-MB-231) compartment was printed onto the adipose tissue constructs. After nine days of co-culture, we observed a cancer cell-induced reduction of the lipid content and a remodeling of the ECM within the adipose tissues, with increased fibronectin, collagen I and collagen VI expression. Together, our data demonstrate that 3D-printed breast cancer-adipose tissue models can recapitulate important aspects of the complex cell–cell and cell–matrix interplay within the tumor-stroma microenvironment.
Collapse
Affiliation(s)
- Hannes Horder
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, 97080 Würzburg, Germany; (H.H.); (M.G.L.); (P.B.-K.)
| | - Mar Guaza Lasheras
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, 97080 Würzburg, Germany; (H.H.); (M.G.L.); (P.B.-K.)
| | - Nadine Grummel
- Department of Physics, Friedrich-Alexander University Erlangen-Nürnberg, 91052 Erlangen, Germany; (N.G.); (B.F.)
| | - Ali Nadernezhad
- Chair for Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, 97080 Würzburg, Germany; (A.N.); (J.H.); (J.T.); (J.G.)
| | - Johannes Herbig
- Chair for Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, 97080 Würzburg, Germany; (A.N.); (J.H.); (J.T.); (J.G.)
| | - Süleyman Ergün
- Department of Medicine, Institute of Anatomy and Cell Biology, University of Würzburg, 97070 Würzburg, Germany;
| | - Jörg Teßmar
- Chair for Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, 97080 Würzburg, Germany; (A.N.); (J.H.); (J.T.); (J.G.)
| | - Jürgen Groll
- Chair for Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, 97080 Würzburg, Germany; (A.N.); (J.H.); (J.T.); (J.G.)
| | - Ben Fabry
- Department of Physics, Friedrich-Alexander University Erlangen-Nürnberg, 91052 Erlangen, Germany; (N.G.); (B.F.)
| | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, 97080 Würzburg, Germany; (H.H.); (M.G.L.); (P.B.-K.)
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, 97080 Würzburg, Germany; (H.H.); (M.G.L.); (P.B.-K.)
- Correspondence: ; Tel.: +49-931-201-37115
| |
Collapse
|
7
|
Ng S, Kurisawa M. Integrating biomaterials and food biopolymers for cultured meat production. Acta Biomater 2021; 124:108-129. [PMID: 33472103 DOI: 10.1016/j.actbio.2021.01.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Cultured meat has recently achieved mainstream prominence due to the emergence of societal and industrial interest. In contrast to animal-based production of traditional meat, the cultured meat approach entails laboratory cultivation of engineered muscle tissue. However, bioengineers have hitherto engineered tissues to fulfil biomedical endpoints, and have had limited experience in engineering muscle tissue for its post-mortem traits, which broadly govern consumer definitions of meat quality. Furthermore, existing tissue engineering approaches face fundamental challenges in technical feasibility and industrial scalability for cultured meat production. This review discusses how animal-based meat production variables influence meat properties at both the molecular and functional level, and whether current cultured meat approaches recapitulate these properties. In addition, this review considers how conventional meat producers employ exogenous biopolymer-based meat ingredients and processing techniques to mimic desirable meat properties in meat products. Finally, current biomaterial strategies for engineering muscle and adipose tissue are surveyed in the context of emerging constraints that pertain to cultured meat production, such as edibility, sustainability and scalability, and potential areas for integrating biomaterials and food biopolymer approaches to address these constraints are discussed. STATEMENT OF SIGNIFICANCE: Laboratory-grown or cultured meat has gained increasing interest from industry and the public, but currently faces significant impediment to market feasibility. This is due to fundamental knowledge gaps in producing realistic meat tissues via conventional tissue engineering approaches, as well as translational challenges in scaling up these approaches in an efficient, sustainable and high-volume manner. By defining the molecular basis for desirable meat quality attributes, such as taste and texture, and introducing the fundamental roles of food biopolymers in mimicking these properties in conventional meat products, this review aims to bridge the historically disparate fields of meat science and biomaterials engineering in order to inspire potentially synergistic strategies that address some of these challenges.
Collapse
|
8
|
Silva DDD, Paz AHDR, Portinho CP, Lima EOC, Kliemann LM, Collares MVM. Reconstruction of parietal bone defects with adiposederived mesenchymal stem cells. Experimental study. Acta Cir Bras 2021; 35:e351201. [PMID: 33503214 PMCID: PMC7819692 DOI: 10.1590/acb351201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/09/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose: This study assessed the regeneration potential of mesenchymal stem cells
(MSC) from adipose tissue associated with platelet-rich plasma (PRP) in bone
regeneration. Methods: Thirty Wistar rats (Rattus norvegicus albinos) were divided into five groups
(according to the grafting material and time to euthanasia): (1) autograft -
14 days (control), (2) autograft - 28 days (control), (3) MSC + PRP - 14
days, (4) MSC + PRP + papaverine - 14 days and (5) MSC + PRP + papaverine -
28 days. After euthanasia, the graft was removed and histological slides
were prepared. They were assessed by a blinded pathologist using a
previously published histological scale as parameter. Results: There was some degree of neoformed bone trabeculae (NBT) in 93.3% of the
samples, as well as osteoblastic activity (OA). The autograft groups (14 and
28 days) had higher levels in the formation of bone trabeculae.
Nonparametric data were analyzed using the Wilcoxon-Mann-Whitney test and
proved not to be statistically significant at p < 0.05. Conclusions: Experimental parietal bone reconstruction, combining MSC, PRP and papaverine
presented regeneration in all groups with no significant difference among
them.
Collapse
|
9
|
Frazier T, Alarcon A, Wu X, Mohiuddin OA, Motherwell JM, Carlsson AH, Christy RJ, Edwards JV, Mackin RT, Prevost N, Gloster E, Zhang Q, Wang G, Hayes DJ, Gimble JM. Clinical Translational Potential in Skin Wound Regeneration for Adipose-Derived, Blood-Derived, and Cellulose Materials: Cells, Exosomes, and Hydrogels. Biomolecules 2020; 10:E1373. [PMID: 32992554 PMCID: PMC7650547 DOI: 10.3390/biom10101373] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Acute and chronic skin wounds due to burns, pressure injuries, and trauma represent a substantial challenge to healthcare delivery with particular impacts on geriatric, paraplegic, and quadriplegic demographics worldwide. Nevertheless, the current standard of care relies extensively on preventive measures to mitigate pressure injury, surgical debridement, skin flap procedures, and negative pressure wound vacuum measures. This article highlights the potential of adipose-, blood-, and cellulose-derived products (cells, decellularized matrices and scaffolds, and exosome and secretome factors) as a means to address this unmet medical need. The current status of this research area is evaluated and discussed in the context of promising avenues for future discovery.
Collapse
Affiliation(s)
- Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Andrea Alarcon
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| | - Omair A. Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi 75270, Pakistan;
| | | | - Anders H. Carlsson
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Robert J. Christy
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, San Antonio, TX 78234, USA; (A.H.C.); (R.J.C.)
| | - Judson V. Edwards
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Robert T. Mackin
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Nicolette Prevost
- Southern Regional Research Center-USDA-ARS, New Orleans, LA 70124, USA; (J.V.E.); (R.T.M.); (N.P.)
| | - Elena Gloster
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Qiang Zhang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA; (E.G.); (Q.Z.); (G.W.)
| | - Daniel J. Hayes
- Department of Biomedical Engineering, State College, Pennsylvania State University, Centre County, PA 16802, USA;
| | - Jeffrey M. Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (A.A.); (X.W.)
- LaCell LLC, New Orleans, LA 70148, USA
| |
Collapse
|
10
|
Wang G, Qiu L, Li X, Pan Y, Sheng Y, Neumann K, Sun Y, Wang Y, Liu L, Deng L, Bradley M, Zhang R. Novel copolymers drive differentiation of human adipose derived stem cells towards chondrocytes and osteoblasts identified by high-throughput approach. Biomed Phys Eng Express 2020; 6:025005. [PMID: 33438631 DOI: 10.1088/2057-1976/ab7155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Human adipose derived stem cells (hASCs) were seeded onto polymer microarrays that had been fabricated using a variety of acrylate monomers to discover novel substrates that induced differentiation towards chondrocytes and osteoblasts. Flow cytometric analysis showed that both CD105 and CD49d positive hASCs increased rapidly with passage number on the lead polymers, while quantitative PCR analysis showed that the substrate synthesized from methacryloxyethyltrimethyl ammonium chloride, N,N-diethylaminoethyl methacrylate and cyclohexyl methacrylate enhanced chondrogenesis and osteogenensis some 4 and 25 times respectively in terms of the expression of SOX9 and ALP in differentiated stem cells. These copolymers substrates thus have great potential for application in the purification, generation and expansion of defined hASC's and the controlled differentiation of of cells for possible clinical application.
Collapse
Affiliation(s)
- Guirong Wang
- Jiangsu Key Laboratory of Environmentally Friendly Polymeric Materials, School of Materials Science and Engineering, Changzhou University, Changzhou 213164, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
12
|
Decoration of RGD-mimetic porous scaffolds with engineered and devitalized extracellular matrix for adipose tissue regeneration. Acta Biomater 2018; 73:154-166. [PMID: 29684623 DOI: 10.1016/j.actbio.2018.04.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/22/2018] [Accepted: 04/19/2018] [Indexed: 12/16/2022]
Abstract
Fat grafting is emerging as a promising alternative to silicon implants in breast reconstruction surgery. Unfortunately, this approach does not provide a proper mechanical support and is affected by drawbacks such as tissue resorption and donor site morbidity. Synthetic scaffolds can offer a valuable alternative to address these challenges, but poorly recapitulate the biochemical stimuli needed for tissue regeneration. Here, we aim at combining the positive features of a structural, synthetic polymer to an engineered, devitalized extracellular matrix (ECM) to generate a hybrid construct that can provide a mix of structural and biological stimuli needed for adipose tissue regeneration. A RGD-mimetic synthetic scaffold OPAAF, designed for soft tissue engineering, was decorated with ECM deposited by human adipose stromal cells (hASCs). The adipoinductive potential of the hybrid ECM-OPAAF construct was validated in vitro, by culture with hASC in a perfusion bioreactor system, and in vivo, by subcutaneous implantation in nude mouse. Our findings demonstrate that the hybrid ECM-OPAAF provides proper mechanical support and adipoinductive stimuli, with potential applicability as off-the-shelf material for adipose tissue reconstruction. STATEMENT OF SIGNIFICANCE In this study we combined the functionalities of a synthetic polymer with those of an engineered and subsequently devitalized extracellular matrix (ECM) to generate a hybrid material for adipose tissue regeneration. The developed hybrid ECM-OPAAF was demonstrated to regulate human adipose stromal cells adipogenic commitment in vitro and adipose tissue infiltration in vivo. Our findings demonstrate that the hybrid ECM-OPAAF provide proper mechanical support and adipoinductive stimuli and represents a promising off-the-shelf material for adipose tissue reconstruction. We believe that our approach could offer an alternative strategy for adipose tissue reconstruction in case of mastectomy or congenital abnormalities, overcoming the current limitations of autologous fat based strategies such as volume resorption and donor site morbidity.
Collapse
|
13
|
Zhu Y, Kruglikov IL, Akgul Y, Scherer PE. Hyaluronan in adipogenesis, adipose tissue physiology and systemic metabolism. Matrix Biol 2018; 78-79:284-291. [PMID: 29458140 PMCID: PMC6534160 DOI: 10.1016/j.matbio.2018.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 02/07/2023]
Abstract
Hyaluronic acid (HA, also known as hyaluronan), is a non-sulfated linear glycosaminoglycan polymer consisting of repeating disaccharide units of d-glucuronic acid and N-acetyl-d-glucosamine abundantly present in the extracellular matrix. The sizes of hyaluronic acid polymers range from 5000 to 20,000,000 Da in vivo, and the functions of HA are largely dictated by its size. Due to its high biocompatibility, HA has been commonly used as soft tissue filler as well as a major component of biomaterial scaffolds in tissue engineering. Several studies have implicated that HA may promote differentiation of adipose tissue derived stem cells in vitro or in vivo when used as a supporting scaffold. However, whether HA actually promotes adipogenesis in vivo and the subsequent metabolic effects of this process are unclear. This review summarizes some recent publications in the field and discusses the possible directions and approaches for future studies, focusing on the role of HA in the adipose tissue.
Collapse
Affiliation(s)
- Yi Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Yucel Akgul
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Aurora A, Wrice N, Walters TJ, Christy RJ, Natesan S. A PEGylated platelet free plasma hydrogel based composite scaffold enables stable vascularization and targeted cell delivery for volumetric muscle loss. Acta Biomater 2018; 65:150-162. [PMID: 29128541 DOI: 10.1016/j.actbio.2017.11.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 10/26/2017] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix (ECM) scaffolds are being used for the clinical repair of soft tissue injuries. Although improved functional outcomes have been reported, ECM scaffolds show limited tissue specific remodeling response with concomitant deposition of fibrotic tissue. One plausible explanation is the regression of blood vessels which may be limiting the diffusion of oxygen and nutrients across the scaffold. Herein we develop a composite scaffold as a vasculo-inductive platform by integrating PEGylated platelet free plasma (PFP) hydrogel with a muscle derived ECM scaffold (m-ECM). In vitro, adipose derived stem cells (ASCs) seeded onto the composite scaffold differentiated into two distinct morphologies, a tubular network in the hydrogel, and elongated structures along the m-ECM scaffold. The composite scaffold showed a high expression of ITGA5, ITGB1, and FN and a synergistic up-regulation of ang1 and tie-2 transcripts. The in vitro ability of the composite scaffold to provide extracellular milieu for cell adhesion and molecular cues to support vessel formation was investigated in a rodent volumetric muscle loss (VML) model. The composite scaffold delivered with ASCs supported robust and stable vascularization. Additionally, the composite scaffold supported increased localization of ASCs in the defect demonstrating its ability for localized cell delivery. Interestingly, ASCs were observed homing in the injured muscle and around the perivascular space possibly to stabilize the host vasculature. In conclusion, the composite scaffold delivered with ASCs presents a promising approach for scaffold vascularization. The versatile nature of the composite scaffold also makes it easily adaptable for the repair of soft tissue injuries. STATEMENT OF SIGNIFICANCE Decellularized extracellular matrix (ECM) scaffolds when used for soft tissue repair is often accompanied by deposition of fibrotic tissue possibly due to limited scaffold vascularization, which limits the diffusion of oxygen and nutrients across the scaffold. Although a variety of scaffold vascularization strategies has been investigated, their limitations preclude rapid clinical translation. In this study we have developed a composite scaffold by integrating bi-functional polyethylene glycol modified platelet free plasma (PEGylated PFP) with adipose derived stem cells (ASCs) along with a muscle derived ECM scaffold (m-ECM). The composite scaffold provides a vasculo-inductive and an effective cell delivery platform for volumetric muscle loss.
Collapse
|
15
|
Decellularized Adipose Tissue Scaffolds for Soft Tissue Regeneration and Adipose-Derived Stem/Stromal Cell Delivery. Methods Mol Biol 2018; 1773:53-71. [PMID: 29687381 DOI: 10.1007/978-1-4939-7799-4_6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Surgically discarded adipose tissue is not only an abundant source of multipotent adipose-derived stem/stromal cells (ASCs) but can also be decellularized to obtain a biomimetic microenvironment for tissue engineering applications. The decellularization methods involve processing excised fat through a series of chemical, mechanical, and enzymatic treatment stages designed to extract cells, cellular components, and lipid from the tissues. This process yields a complex 3D bioscaffold enriched in collagens that mimics the biochemical and biomechanical properties of the native extracellular matrix (ECM). For ASC culture and delivery, decellularized adipose tissue (DAT) provides a cell-supportive platform that is conducive to adipogenesis. While DAT can be applied in its intact form as an off-the-shelf adipogenic matrix, it can also be used as an ECM source for the fabrication of an array of other scaffold formats including adipose ECM-derived microcarriers and porous foams. In this chapter, we describe the methods developed in our lab to decellularize human adipose tissue and to further process it into a variety of scaffolding materials for a range of applications in soft tissue regeneration, wound healing, and cell culture.
Collapse
|
16
|
Kessler L, Gehrke S, Winnefeld M, Huber B, Hoch E, Walter T, Wyrwa R, Schnabelrauch M, Schmidt M, Kückelhaus M, Lehnhardt M, Hirsch T, Jacobsen F. Methacrylated gelatin/hyaluronan-based hydrogels for soft tissue engineering. J Tissue Eng 2017; 8:2041731417744157. [PMID: 29318000 PMCID: PMC5753891 DOI: 10.1177/2041731417744157] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022] Open
Abstract
In vitro–generated soft tissue could provide alternate therapies for soft tissue defects. The aim of this study was to evaluate methacrylated gelatin/hyaluronan as scaffolds for soft tissue engineering and their interaction with human adipose–derived stem cells (hASCs). ASCs were incorporated into methacrylated gelatin/hyaluronan hydrogels. The gels were photocrosslinked with a lithium phenyl-2,4,6-trimethylbenzoylphosphinate photoinitiator and analyzed for cell viability and adipogenic differentiation of ASCs over a period of 30 days. Additionally, an angiogenesis assay was performed to assess their angiogenic potential. After 24 h, ASCs showed increased viability on composite hydrogels. These results were consistent over 21 days of culture. By induction of adipogenic differentiation, the mature adipocytes were observed after 7 days of culture, their number significantly increased until day 28 as well as expression of fatty acid binding protein 4 and adiponectin. Our scaffolds are promising as building blocks for adipose tissue engineering and allowed long viability, proliferation, and differentiation of ASCs.
Collapse
Affiliation(s)
- Lukas Kessler
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| | - Sandra Gehrke
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Marc Winnefeld
- Research and Development, Beiersdorf AG, Hamburg, Germany
| | - Birgit Huber
- Institute for Interfacial Engineering and Plasma Technology, University of Stuttgart, Stuttgart, Germany
| | - Eva Hoch
- Institute for Interfacial Engineering and Plasma Technology, University of Stuttgart, Stuttgart, Germany
| | | | - Ralf Wyrwa
- Biomaterials Department, INNOVENT e. V., Jena, Germany
| | | | - Malte Schmidt
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| | - Maximilian Kückelhaus
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| | - Tobias Hirsch
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| | - Frank Jacobsen
- Department of Plastic Surgery and Burn Centre, BG University Hospital Bergmannsheil GmbH, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
17
|
Wang X, Chen G, Huang C, Tu H, Zou J, Yan J. Bone marrow stem cells-derived extracellular matrix is a promising material. Oncotarget 2017; 8:98336-98347. [PMID: 29228693 PMCID: PMC5716733 DOI: 10.18632/oncotarget.21683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 09/20/2017] [Indexed: 01/08/2023] Open
Abstract
The extracellular matrix(ECM), which is primarily composed of collagens and proteoglycans, plays a key role in cell proliferation, differentiation, and migration and interactions between cells. In this study, we produced chitosan/gelatin/bone marrow stem cells-derived extracellular matrix(C/G/BMSCs-dECM) scaffolds via lyophilization and cross-linking, and chitosan/gelatin(C/G) scaffolds were used as controls. For the C/G/BMSCs-dECM scaffolds, the average pore size was 289.17 ± 80.28 μm; the average porosity was 89.25 ± 3.75%; the average compressive modulus was 0.82 ± 0.07 MPa; and the average water uptake ratio was 13.81 ± 1.00. In vitro, the C/G/BMSCs-dECM scaffolds promoted bone marrow stem cells(BMSCs) attachment and proliferation. Moreover, improved osteogenic differentiation was observed for these scaffolds. Thus, C/G/BMSCs-dECM is a promising material for bone tissue engineering.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P.R. China.,Key Laboratory of the Ministry of Education, Ministry of Myocardial Ischemia, Harbin 150086, P.R. China
| | - Guanghua Chen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P.R. China.,Key Laboratory of the Ministry of Education, Ministry of Myocardial Ischemia, Harbin 150086, P.R. China
| | - Chao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P.R. China
| | - Hualei Tu
- Burn Department, The Fifth Hospital of Harbin, Harbin 150086, P.R. China
| | - Jilong Zou
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P.R. China
| |
Collapse
|
18
|
Characterization of human adipose tissue-derived stem cells in vitro culture and in vivo differentiation in a temperature-sensitive chitosan/β- glycerophosphate/collagen hybrid hydrogel. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:231-240. [DOI: 10.1016/j.msec.2016.08.085] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 07/29/2016] [Accepted: 08/30/2016] [Indexed: 11/17/2022]
|
19
|
Composite Bioscaffolds Incorporating Decellularized ECM as a Cell-Instructive Component Within Hydrogels as In Vitro Models and Cell Delivery Systems. Methods Mol Biol 2017; 1577:183-208. [PMID: 28493212 DOI: 10.1007/7651_2017_36] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decellularized tissues represent promising biomaterials, which harness the innate capacity of the tissue-specific extracellular matrix (ECM) to direct cell functions including stem cell proliferation and lineage-specific differentiation. However, bioscaffolds derived exclusively from decellularized ECM offer limited versatility in terms of tuning biomechanical properties, as well as cell-cell and cell-ECM interactions that are important mediators of the cellular response. As an alternative approach, in the current chapter we describe methods for incorporating cryo-milled decellularized tissues as a cell-instructive component within a hydrogel carrier designed to crosslink under mild conditions. This composite strategy can enable in situ cell encapsulation with high cell viability, allowing efficient seeding with a homogeneous distribution of cells and ECM. Detailed protocols are provided for the effective decellularization of human adipose tissue and porcine auricular cartilage, as well as the cryo-milling process used to generate the ECM particles. Further, we describe methods for synthesizing methacrylated chondroitin sulphate (MCS) and for performing UV-initiated and thermally induced crosslinking to form hydrogel carriers for adipose and cartilage regeneration. The hydrogel composites offer great flexibility, and the hydrogel phase, ECM source, particle size, cell type(s) and seeding density can be tuned to promote the desired cellular response. Overall, these systems represent promising platforms for the development of tissue-specific 3-D in vitro cell culture models and in vivo cell delivery systems.
Collapse
|
20
|
Decellularized skin/adipose tissue flap matrix for engineering vascularized composite soft tissue flaps. Acta Biomater 2016; 35:166-84. [PMID: 26876876 DOI: 10.1016/j.actbio.2016.02.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 02/06/2016] [Accepted: 02/11/2016] [Indexed: 02/07/2023]
Abstract
Using a perfusion decellularization protocol, we developed a decellularized skin/adipose tissue flap (DSAF) comprising extracellular matrix (ECM) and intact vasculature. Our DSAF had a dominant vascular pedicle, microcirculatory vascularity, and a sensory nerve network and retained three-dimensional (3D) nanofibrous structures well. DSAF, which was composed of collagen and laminin with well-preserved growth factors (e.g., vascular endothelial growth factor, basic fibroblast growth factor), was successfully repopulated with human adipose-derived stem cells (hASCs) and human umbilical vein endothelial cells (HUVECs), which integrated with DSAF and formed 3D aggregates and vessel-like structures in vitro. We used microsurgery techniques to re-anastomose the recellularized DSAF into nude rats. In vivo, the engineered flap construct underwent neovascularization and constructive remodeling, which was characterized by the predominant infiltration of M2 macrophages and significant adipose tissue formation at 3months postoperatively. Our results indicate that DSAF co-cultured with hASCs and HUVECs is a promising platform for vascularized soft tissue flap engineering. This platform is not limited by the flap size, as the entire construct can be immediately perfused by the recellularized vascular network following simple re-integration into the host using conventional microsurgical techniques. STATEMENT OF SIGNIFICANCE Significant soft tissue loss resulting from traumatic injury or tumor resection often requires surgical reconstruction using autologous soft tissue flaps. However, the limited availability of qualitative autologous flaps as well as the donor site morbidity significantly limits this approach. Engineered soft tissue flap grafts may offer a clinically relevant alternative to the autologous flap tissue. In this study, we engineered vascularized soft tissue free flap by using skin/adipose flap extracellular matrix scaffold (DSAF) in combination with multiple types of human cells. Following vascular reanastomosis in the recipient site, the engineered products successful regenerated large-scale fat tissue in vivo. This approach may provide a translatable platform for composite soft tissue free flap engineering for microsurgical reconstruction.
Collapse
|
21
|
Calabrese R, Raia N, Huang W, Ghezzi CE, Simon M, Staii C, Weiss AS, Kaplan DL. Silk-ionomer and silk-tropoelastin hydrogels as charged three-dimensional culture platforms for the regulation of hMSC response. J Tissue Eng Regen Med 2016; 11:2549-2564. [DOI: 10.1002/term.2152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/19/2015] [Accepted: 12/22/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Rossella Calabrese
- Department of Biomedical Engineering; Tufts University Science and Technology Center; Medford MA USA
| | - Nicole Raia
- Department of Biomedical Engineering; Tufts University Science and Technology Center; Medford MA USA
| | - Wenwen Huang
- Department of Biomedical Engineering; Tufts University Science and Technology Center; Medford MA USA
| | - Chiara E. Ghezzi
- Department of Biomedical Engineering; Tufts University Science and Technology Center; Medford MA USA
| | - Marc Simon
- Department of Physics and Astronomy, and Center for Nanoscopic Physics; Tufts University Science and Technology Center; Medford MA USA
| | - Cristian Staii
- Department of Physics and Astronomy, and Center for Nanoscopic Physics; Tufts University Science and Technology Center; Medford MA USA
| | - Anthony S. Weiss
- School of Molecular Bioscience; University of Sydney; NSW Australia
- Charles Perkins Center; University of Sydney; NSW Australia
- Bosch Institute; University of Sydney; NSW Australia
| | - David L. Kaplan
- Department of Biomedical Engineering; Tufts University Science and Technology Center; Medford MA USA
| |
Collapse
|
22
|
Kruglikov IL, Wollina U. Soft tissue fillers as non-specific modulators of adipogenesis: change of the paradigm? Exp Dermatol 2015; 24:912-5. [PMID: 26309229 DOI: 10.1111/exd.12852] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 12/15/2022]
Abstract
Dermal filler injection is a cornerstone of facial rejuvenation procedures. Based on available data in animal and human studies, we suppose that the activation and proliferation of adipose-derived stem cells and expansion of mature adipocytes play a crucial role in long-term effects of volumizing, tissue tightening and beautification.
Collapse
Affiliation(s)
| | - Uwe Wollina
- Hospital Dresden-Friedrichstadt, Academic Teaching Hospital of the Technical University of Dresden, Dresden, Germany
| |
Collapse
|
23
|
Henriksson HB, Papadimitriou N, Tschernitz S, Svala E, Skioldebrand E, Windahl S, Junevik K, Brisby H. Indications of that migration of stem cells is influenced by the extra cellular matrix architecture in the mammalian intervertebral disk region. Tissue Cell 2015; 47:439-55. [DOI: 10.1016/j.tice.2015.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 07/30/2015] [Accepted: 08/04/2015] [Indexed: 01/07/2023]
|
24
|
Role of c-Jun N-terminal kinase in the osteogenic and adipogenic differentiation of human adipose-derived mesenchymal stem cells. Exp Cell Res 2015; 339:112-21. [PMID: 26272544 DOI: 10.1016/j.yexcr.2015.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/08/2015] [Accepted: 08/09/2015] [Indexed: 01/02/2023]
Abstract
Although previous studies have characterized the osteogenic potential of adipose-derived mesenchymal stem cells (AMSCs) in vitro and in vivo, the molecular mechanism involved remains to be fully determined. Previously, we demonstrated that the ERK pathway plays an important role in osteogenesis and regulation of the balance between osteogenesis and adipogenesis. Here, we explored the possible role of JNKs in osteogenesis and adipogenesis of AMSCs. JNK activation in osteo-induced AMSCs was initiated at 15 min, peaked at 30 min, and declined from 45 min to basal levels. Inhibition of the JNK signaling pathway using SP600125 blocked osteogenic differentiation in a dose-dependent manner, which was revealed by an ALP activity assay, extracellular calcium deposition detection, and expression of osteogenesis-relative genes (Runx2, ALP, and OCN) via RT-PCR and real-time PCR. However, blockage of JNK did not induce a switch between osteogenesis and adipogenesis of AMSCs in the presence of dexamethasone, which is different from that of blockage of ERK. Significantly, the blockage of JNK activation in adipo-induced AMSCs by SP600125 stimulated adipogenic differentiation, which was confirmed by Oil Red O staining to detect intracellular lipid droplets, and RT-PCR and real-time PCR analysis for expression of adipogenesis-relative genes (PPARγ2 and aP2). This study suggested a potential function of the JNK pathway in committing osteogenic and adipogenic differentiation of AMSCs in vitro. However, blockage of the JNK pathway is not sufficient to induce a switch from osteogenesis to adipogenesis of AMSCs.
Collapse
|
25
|
Suhaeri M, Subbiah R, Van SY, Du P, Kim IG, Lee K, Park K. Cardiomyoblast (h9c2) differentiation on tunable extracellular matrix microenvironment. Tissue Eng Part A 2015; 21:1940-51. [PMID: 25836924 DOI: 10.1089/ten.tea.2014.0591] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrices (ECM) obtained from in vitro-cultured cells have been given much attention, but its application in cardiac tissue engineering is still limited. This study investigates cardiomyogenic potential of fibroblast-derived matrix (FDM) as a novel ECM platform over gelatin or fibronectin, in generating cardiac cell lineages derived from H9c2 cardiomyoblasts. As characterized through SEM and AFM, FDM exhibits unique surface texture and biomechanical property. Immunofluorescence also found fibronectin, collagen, and laminin in the FDM. Cells on FDM showed a more circular shape and slightly less proliferation in a growth medium. After being cultured in a differentiation medium for 7 days, H9c2 cells on FDM differentiated into cardiomyocytes, as identified by stronger positive markers, such as α-actinin and cTnT, along with more elevated gene expression of Myl2 and Tnnt compared to the cells on gelatin and fibronectin. The gap junction protein connexin 43 was also significantly upregulated for the cells differentiated on FDM. A successive work enabled matrix stiffness tunable; FDM crosslinked by 2wt% genipin increased the stiffness up to 8.5 kPa, 100 times harder than that of natural FDM. The gene expression of integrin subunit α5 was significantly more upregulated on FDM than on crosslinked FDM (X-FDM), whereas no difference was observed for β1 expression. Interestingly, X-FDM showed a much greater effect on the cardiomyoblast differentiation into cardiomyocytes over natural one. This study strongly indicates that FDM can be a favorable ECM microenvironment for cardiomyogenesis of H9c2 and that tunable mechanical compliance induced by crosslinking further provides a valuable insight into the role of matrix stiffness on cardiomyogenesis.
Collapse
Affiliation(s)
- Muhammad Suhaeri
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Ramesh Subbiah
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Se Young Van
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Ping Du
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - In Gul Kim
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kangwon Lee
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| | - Kwideok Park
- 1Center for Biomaterials, Korea Institute of Science and Technology, Seoul, Republic of Korea.,2Department of Biomedical Engineering, University of Science and Technology, Daejon, Republic of Korea
| |
Collapse
|
26
|
Wollina U. Midfacial rejuvenation by hyaluronic acid fillers and subcutaneous adipose tissue – A new concept. Med Hypotheses 2015; 84:327-30. [DOI: 10.1016/j.mehy.2015.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 11/27/2022]
|
27
|
Ornamenting 3D printed scaffolds with cell-laid extracellular matrix for bone tissue regeneration. Biomaterials 2014; 37:230-41. [PMID: 25453953 DOI: 10.1016/j.biomaterials.2014.10.012] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/02/2014] [Indexed: 12/31/2022]
Abstract
3D printing technique is the most sophisticated technique to produce scaffolds with tailorable physical properties. But, these scaffolds often suffer from limited biological functionality as they are typically made from synthetic materials. Cell-laid mineralized ECM was shown to be potential for improving the cellular responses and drive osteogenesis of stem cells. Here, we intend to improve the biological functionality of 3D-printed synthetic scaffolds by ornamenting them with cell-laid mineralized extracellular matrix (ECM) that mimics a bony microenvironment. We developed bone graft substitutes by using 3D printed scaffolds made from a composite of polycaprolactone (PCL), poly(lactic-co-glycolic acid) (PLGA), and β-tricalcium phosphate (β-TCP) and mineralized ECM laid by human nasal inferior turbinate tissue-derived mesenchymal stromal cells (hTMSCs). A rotary flask bioreactor was used to culture hTMSCs on the scaffolds to foster formation of mineralized ECM. A freeze/thaw cycle in hypotonic buffer was used to efficiently decellularize (97% DNA reduction) the ECM-ornamented scaffolds while preserving its main organic and inorganic components. The ECM-ornamented 3D printed scaffolds supported osteoblastic differentiation of newly-seeded hTMSCs by upregulating four typical osteoblastic genes (4-fold higher RUNX2; 3-fold higher ALP; 4-fold higher osteocalcin; and 4-fold higher osteopontin) and increasing calcium deposition compared to bare 3D printed scaffolds. In vivo, in ectopic and orthotopic models in rats, ECM-ornamented scaffolds induced greater bone formation than that of bare scaffolds. These results suggest a valuable method to produce ECM-ornamented 3D printed scaffolds as off-the-shelf bone graft substitutes that combine tunable physical properties with physiological presentation of biological signals.
Collapse
|
28
|
Krontiras P, Gatenholm P, Hägg DA. Adipogenic differentiation of stem cells in three-dimensional porous bacterial nanocellulose scaffolds. J Biomed Mater Res B Appl Biomater 2014; 103:195-203. [DOI: 10.1002/jbm.b.33198] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/26/2014] [Accepted: 04/24/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Panagiotis Krontiras
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| | - Paul Gatenholm
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| | - Daniel A Hägg
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| |
Collapse
|
29
|
Tanzi MC, Farè S. Adipose tissue engineering: state of the art, recent advances and innovative approaches. Expert Rev Med Devices 2014; 6:533-51. [DOI: 10.1586/erd.09.37] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Wu I, Elisseeff J. Biomaterials and Tissue Engineering for Soft Tissue Reconstruction. NATURAL AND SYNTHETIC BIOMEDICAL POLYMERS 2014:235-241. [DOI: 10.1016/b978-0-12-396983-5.00015-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
31
|
Liu Y, Fox V, Lei Y, Hu B, Joo KI, Wang P. Synthetic niches for differentiation of human embryonic stem cells bypassing embryoid body formation. J Biomed Mater Res B Appl Biomater 2013; 102:1101-12. [DOI: 10.1002/jbm.b.33092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/16/2013] [Accepted: 11/26/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science; University of Southern California; Los Angeles California
| | - Victoria Fox
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research; University of Southern California; Los Angeles California
| | - Yuning Lei
- Mork Family Department of Chemical Engineering and Materials Science; University of Southern California; Los Angeles California
| | - Biliang Hu
- Mork Family Department of Chemical Engineering and Materials Science; University of Southern California; Los Angeles California
| | - Kye-Il Joo
- Mork Family Department of Chemical Engineering and Materials Science; University of Southern California; Los Angeles California
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science; University of Southern California; Los Angeles California
- Department of Biomedical Engineering; University of Southern California; Los Angeles California
- Department of Pharmacology and Pharmaceutical Sciences; University of Southern California; Los Angeles California
| |
Collapse
|
32
|
Cheung HK, Han TTY, Marecak DM, Watkins JF, Amsden BG, Flynn LE. Composite hydrogel scaffolds incorporating decellularized adipose tissue for soft tissue engineering with adipose-derived stem cells. Biomaterials 2013; 35:1914-23. [PMID: 24331712 DOI: 10.1016/j.biomaterials.2013.11.067] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022]
Abstract
An injectable tissue-engineered adipose substitute that could be used to deliver adipose-derived stem cells (ASCs), filling irregular defects and stimulating natural soft tissue regeneration, would have significant value in plastic and reconstructive surgery. With this focus, the primary aim of the current study was to characterize the response of human ASCs encapsulated within three-dimensional bioscaffolds incorporating decellularized adipose tissue (DAT) as a bioactive matrix within photo-cross-linkable methacrylated glycol chitosan (MGC) or methacrylated chondroitin sulphate (MCS) delivery vehicles. Stable MGC- and MCS-based composite scaffolds were fabricated containing up to 5 wt% cryomilled DAT through initiation with long-wavelength ultraviolet light. The encapsulation strategy allows for tuning of the 3-D microenvironment and provides an effective method of cell delivery with high seeding efficiency and uniformity, which could be adapted as a minimally-invasive in situ approach. Through in vitro cell culture studies, human ASCs were assessed over 14 days in terms of viability, glycerol-3-phosphate dehydrogenase (GPDH) enzyme activity, adipogenic gene expression and intracellular lipid accumulation. In all of the composites, the DAT functioned as a cell-supportive matrix that enhanced ASC viability, retention and adipogenesis within the gels. The choice of hydrogel also influenced the cell response, with significantly higher viability and adipogenic differentiation observed in the MCS composites containing 5 wt% DAT. In vivo analysis in a subcutaneous Wistar rat model at 1, 4 and 12 weeks showed superior implant integration and adipogenesis in the MCS-based composites, with allogenic ASCs promoting cell infiltration, angiogenesis and ultimately, fat formation.
Collapse
Affiliation(s)
- Hoi Ki Cheung
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L 2V7
| | - Tim Tian Y Han
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L 2V7
| | - Dale M Marecak
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L 2V7
| | - John F Watkins
- Department of Surgery, Queen's University, 166 Brock Street, Kingston, Ontario, Canada K7L 5G2
| | - Brian G Amsden
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L 2V7
| | - Lauren E Flynn
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L 2V7; Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, Ontario, Canada K7L 3N6.
| |
Collapse
|
33
|
Wang L, Johnson JA, Zhang Q, Beahm EK. Combining decellularized human adipose tissue extracellular matrix and adipose-derived stem cells for adipose tissue engineering. Acta Biomater 2013; 9:8921-31. [PMID: 23816649 DOI: 10.1016/j.actbio.2013.06.035] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/08/2013] [Accepted: 06/20/2013] [Indexed: 02/08/2023]
Abstract
Repair of soft tissue defects resulting from lumpectomy or mastectomy has become an important rehabilitation process for breast cancer patients. This study aimed to provide an adipose tissue engineering platform for soft tissue defect repair by combining decellularized human adipose tissue extracellular matrix (hDAM) and human adipose-derived stem cells (hASCs). To derive hDAM incised human adipose tissues underwent a decellularization process. Effective cell removal and lipid removal were proved by immunohistochemical analysis and DNA quantification. Scanning electron microscopic examination showed a three-dimensional nanofibrous architecture in hDAM. The hDAM included collagen, sulfated glycosaminoglycan, and vascular endothelial growth factor, but lacked major histocompatibility complex antigen I. hASC viability and proliferation on hDAM were proven in vitro. hDAM implanted subcutaneously in Fischer rats did not cause an immunogenic response, and it underwent remodeling, as indicated by host cell infiltration, neovascularization, and adipose tissue formation. Fresh fat grafts (Coleman technique) and engineered fat grafts (hDAM combined with hASCs) were implanted subcutaneously in nude rats. The implanted engineered fat grafts maintained their volume for 8 weeks, and the hASCs contributed to adipose tissue formation. In summary, the combination of hDAM and hASCs provides not only a clinically translatable platform for adipose tissue engineering, but also a vehicle for elucidating fat grafting mechanisms.
Collapse
|
34
|
Kulig KM, Luo X, Finkelstein EB, Liu XH, Goldman SM, Sundback CA, Vacanti JP, Neville CM. Biologic properties of surgical scaffold materials derived from dermal ECM. Biomaterials 2013; 34:5776-84. [PMID: 23642537 DOI: 10.1016/j.biomaterials.2013.02.055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/19/2013] [Indexed: 01/15/2023]
Abstract
Surgical scaffold materials manufactured from donor human or animal tissue are increasingly being used to promote soft tissue repair and regeneration. The clinical product consists of the residual extracellular matrix remaining after a rigorous decellularization process. Optimally, the material provides both structural support during the repair period and cell guidance cues for effective incorporation into the regenerating tissue. Surgical scaffold materials are available from several companies and are unique products manufactured by proprietary methodology. A significant need exists for a more thorough understanding of scaffold properties that impact the early steps of host cell recruitment and infiltration. In this study, a panel of in vitro assays was used to make direct comparisons of several similar, commercially-available materials: Alloderm, Medeor Matrix, Permacol, and Strattice. Differences in the materials were detected for both cell signaling and scaffold architecture-dependent cell invasion. Material-conditioned media studies found Medeor Matrix to have the greatest positive effect upon cell proliferation and induction of migration. Strattice provided the greatest chemotaxis signaling and best suppressed apoptotic induction. Among assays measuring structure-dependent properties, Medeor Matrix was superior for cell attachment, followed by Permacol. Only Alloderm and Medeor Matrix supported chemotaxis-driven cell invasion beyond the most superficial zone. Medeor Matrix was the only material in the chorioallantoic membrane assay to support substantial cell invasion. These results indicate that both biologic and structural properties need to be carefully assessed in the considerable ongoing efforts to develop new uses and products in this important class of biomaterials.
Collapse
Affiliation(s)
- Katherine M Kulig
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Gimble J, Rad MR, Yao S. Adipose Tissue–Derived Stem Cells and Their Regeneration Potential. STEM CELLS IN CRANIOFACIAL DEVELOPMENT AND REGENERATION 2013:241-258. [DOI: 10.1002/9781118498026.ch12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
36
|
Porzionato A, Sfriso MM, Macchi V, Rambaldo A, Lago G, Lancerotto L, Vindigni V, De Caro R. Decellularized omentum as novel biologic scaffold for reconstructive surgery and regenerative medicine. Eur J Histochem 2013; 57:e4. [PMID: 23549463 PMCID: PMC3683611 DOI: 10.4081/ejh.2013.e4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/17/2012] [Accepted: 09/19/2012] [Indexed: 12/17/2022] Open
Abstract
Homologous tissues, such as adipose tissue, may be an interesting source of acellular scaffolds, maintaining a complex physiological three-dimensional (3D) structure, to be recellularized with autologous cells. The aim of the present work is to evaluate the possibility of obtaining homologous acellular scaffolds from decellularization of the omentum, which is known to have a complex vascular network. Adult rat and human omenta were treated with an adapted decellularization protocol involving mechanical rupture (freeze-thaw cycles), enzymatic digestion (trypsin, lipase, deoxyribonuclease, ribonuclease) and lipid extraction (2-propanol). Histological staining confirmed the effectiveness of decellularization, resulting in cell-free scaffolds with no residual cells in the matrix. The complex 3D networks of collagen (azan-Mallory), elastic fibers (Van Gieson), reticular fibers and glycosaminoglycans (PAS) were maintained, whereas Oil Red and Sudan stains showed the loss of lipids in the decellularized tissue. The vascular structures in the tissue were still visible, with preservation of collagen and elastic wall components and loss of endothelial (anti-CD31 and -CD34 immunohistochemistry) and smooth muscle (anti-alpha smooth muscle actin) cells. Fat-rich and well vascularized omental tissue may be decellularized to obtain complex 3D scaffolds preserving tissue architecture potentially suitable for recellularization. Further analyses are necessary to verify the possibility of recolonization of the scaffold by adipose-derived stem cells in vitro and then in vivo after re implantation, as already known for homologus implants in regenerative processes.
Collapse
Affiliation(s)
- A Porzionato
- Section of Human Anatomy, Department of Molecular Medicine, University of Padua, Padova,
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Adipose tissue pathologies and defects have always represented a reconstructive challenge for plastic surgeons. In more recent years, several allogenic and alloplastic materials have been developed and used as fillers for soft tissue defects. However, their clinical use has been limited by further documented complications, such as foreign-body reactions potentially affecting function, degradation over time, and the risk for immunogenicity. Tissue-engineering strategies are thus being investigated to develop methods for generating adipose tissue. This paper will discuss the current state of the art in adipose tissue engineering techniques, exploring the biomaterials used, stem cells application, culture strategies, and current regulatory framework that are in use are here described and discussed.
Collapse
|
38
|
Flynn L, Woodhouse KA. Adipose tissue engineering with cells in engineered matrices. Organogenesis 2012; 4:228-35. [PMID: 19337402 DOI: 10.4161/org.4.4.7082] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 09/29/2008] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering has shown promise for the development of constructs to facilitate large volume soft tissue augmentation in reconstructive and cosmetic plastic surgery. This article reviews the key progress to date in the field of adipose tissue engineering. In order to effectively design a soft tissue substitute, it is critical to understand the native tissue environment and function. As such, the basic physiology of adipose tissue is described and the process of adipogenesis is discussed. In this article, we have focused on tissue engineering using a cell-seeded scaffold approach, where engineered extracellular matrix substitutes are seeded with exogenous cells that may contribute to the regenerative response. The strengths and limitations of each of the possible cell sources for adipose tissue engineering, including adipose-derived stem cells, are detailed. We briefly highlight some of the results from the major studies to date, involving a range of synthetic and naturally derived scaffolds. While these studies have shown that adipose tissue regeneration is possible, more research is required to develop optimized constructs that will facilitate safe, predictable and long-term augmentation in clinical applications.
Collapse
Affiliation(s)
- Lauren Flynn
- Department of Chemical Engineering; Queen's University; Ontario Canada
| | | |
Collapse
|
39
|
Kang Y, Kim S, Bishop J, Khademhosseini A, Yang Y. The osteogenic differentiation of human bone marrow MSCs on HUVEC-derived ECM and β-TCP scaffold. Biomaterials 2012; 33:6998-7007. [PMID: 22795852 DOI: 10.1016/j.biomaterials.2012.06.061] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 06/22/2012] [Indexed: 01/06/2023]
Abstract
Extracellular matrix (ECM) serves a key role in cell migration, attachment, and cell development. Here we report that ECM derived from human umbilical vein endothelial cells (HUVEC) promoted osteogenic differentiation of human bone marrow mesenchymal stem cells (hMSC). We first produced an HUVEC-derived ECM on a three-dimensional (3D) beta-tricalcium phosphate (β-TCP) scaffold by HUVEC seeding, incubation, and decellularization. The HUVEC-derived ECM was then characterized by SEM, FTIR, XPS, and immunofluorescence staining. The effect of HUVEC-derived ECM-containing β-TCP scaffold on hMSC osteogenic differentiation was subsequently examined. SEM images indicate a dense matrix layer deposited on the surface of struts and pore walls. FTIR and XPS measurements show the presence of new functional groups (amide and hydroxyl groups) and elements (C and N) in the ECM/β-TCP scaffold when compared to the β-TCP scaffold alone. Immunofluorescence images indicate that high levels of fibronectin and collagen IV and low level of laminin were present on the scaffold. ECM-containing β-TCP scaffolds significantly increased alkaline phosphatase (ALP) specific activity and up-regulated expression of osteogenesis-related genes such as runx2, alkaline phosphatase, osteopontin and osteocalcin in hMSC, compared to β-TCP scaffolds alone. This increased effect was due to the activation of MAPK/ERK signaling pathway since disruption of this pathway using an ERK inhibitor PD98059 results in down-regulation of these osteogenic genes. Cell-derived ECM-containing calcium phosphate scaffolds is a promising osteogenic-promoting bone void filler in bone tissue regeneration.
Collapse
Affiliation(s)
- Yunqing Kang
- Department of Orthopedic Surgery, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
40
|
Sukarto A, Yu C, Flynn LE, Amsden BG. Co-delivery of adipose-derived stem cells and growth factor-loaded microspheres in RGD-grafted N-methacrylate glycol chitosan gels for focal chondral repair. Biomacromolecules 2012; 13:2490-502. [PMID: 22746668 DOI: 10.1021/bm300733n] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The coencapsulation of growth factor-loaded microspheres with adipose-derived stem cells (ASCs) within a hydrogel matrix was studied as a potential means to enhance ASC chondrogenesis in the development of a cell-based therapeutic strategy for the regeneration of partial thickness chondral defects. A photopolymerizable N-methacrylate glycol chitosan (MGC) was employed to form an in situ gel used to encapsulate microspheres loaded with bone morphogenetic protein 6 (BMP-6) and transforming growth factor-β3 (TGF-β3) with human ASCs. ASC viability and retention were enhanced when the Young's modulus of the MGC ranged between 225 and 380 kPa. Grafting an RGD-containing peptide onto the MGC backbone (RGD-MGC) improved ASC viability within the gels, remaining at greater than 90% over 14 days in culture. The effects of BMP-6 and TGF-β3 released from the polymer microspheres on ASC chondrogenesis were assessed, and the level of differentiation was compared to ASCs in control gels containing nongrowth factor-loaded microspheres cultured with and without the growth factors supplied in the medium. There was enhanced expression of chondrogenic markers at earlier time points when the ASCs were induced with the sustained and local release of BMP-6 and TGF-β3 from the microspheres. More specifically, the normalized glycosaminoglycan and collagen type II protein expression levels were significantly higher than in the controls. In addition, the ratio of collagen type II to type I was significantly higher in the microsphere delivery group and increased over time. End-point RT-PCR analysis supported that there was a more rapid induction and enhancement of ASC chondrogenesis in the controlled release group. Interestingly, in all of the assays, there was evidence of chondrogenic differentiation when the ASCs were cultured in the gels in the absence of growth factor stimulation. Overall, the co-delivery of growth-factor-loaded microspheres and ASCs in RGD-modified MGC gels successfully induced ASC chondrogenesis and is a promising strategy for cartilage repair.
Collapse
Affiliation(s)
- Abby Sukarto
- Department of Chemical Engineering, Queen's University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
41
|
Lee DW, Jeon YR, Cho EJ, Kang JH, Lew DH. Optimal administration routes for adipose-derived stem cells therapy in ischaemic flaps. J Tissue Eng Regen Med 2012; 8:596-603. [DOI: 10.1002/term.1552] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/18/2012] [Accepted: 05/25/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Dong Won Lee
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration; Yonsei University Health System; Severance Hospital Seoul Republic of Korea
| | - Yeo Reum Jeon
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration; Yonsei University Health System; Severance Hospital Seoul Republic of Korea
| | - Eul Je Cho
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration; Yonsei University Health System; Severance Hospital Seoul Republic of Korea
| | - Jong Hwa Kang
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration; Yonsei University Health System; Severance Hospital Seoul Republic of Korea
| | - Dae Hyun Lew
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration; Yonsei University Health System; Severance Hospital Seoul Republic of Korea
| |
Collapse
|
42
|
Magrofuoco E, Elvassore N, Doyle FJ. Theoretical analysis of insulin-dependent glucose uptake heterogeneity in 3D bioreactor cell culture. Biotechnol Prog 2012; 28:833-45. [DOI: 10.1002/btpr.1539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 02/27/2012] [Indexed: 11/08/2022]
|
43
|
Abstract
Adipose tissue engineering has recently gained significant attention from materials scientists as a result of the exponential growth of soft tissue filler procedures being performed within the clinic. While several injectable materials are currently being marketed for filling subcutaneous voids, they often face limited longevity due to rapid resorption. Their inability to encourage natural adipose formation or ingrowth necessitates repeated injections for a prolonged effect and thus classifies them as temporary fillers. As a result, a significant need for injectable materials that not only act as fillers but also promote in vivo adipogenesis is beginning to be realized. This paper will discuss the advantages and disadvantages of commercially available soft tissue fillers. It will then summarize the current state of research using injectable synthetic materials, biopolymers and extracellular matrix-derived materials for adipose tissue engineering. Furthermore, the successful attributes observed across each of these materials will be outlined along with a discussion of the current difficulties and future directions for adipose tissue engineering.
Collapse
Affiliation(s)
- D A Young
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412, USA
| | | |
Collapse
|
44
|
Monfort A, Soriano-Navarro M, García-Verdugo JM, Izeta A. Production of human tissue-engineered skin trilayer on a plasma-based hypodermis. J Tissue Eng Regen Med 2012; 7:479-90. [PMID: 22294482 DOI: 10.1002/term.548] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 10/18/2011] [Accepted: 11/15/2011] [Indexed: 01/28/2023]
Abstract
Full thickness wounds require a dermal component to achieve functional permanent skin restoration. Currently available tissue-engineered skin substitutes lack a subcutaneous fat layer that would functionally contribute some of the mechanical and thermoregulatory properties of normal skin. To generate a trilayer engineered skin equivalent, we included bone marrow mesenchymal (BM-MSC) or adipose tissue-derived (ASC) stromal cells in a human plasma hydrogel exposed to adipogenic clues for three weeks. Approximately half of the cells differentiated under these conditions into mature adipocytes that survived for two years in culture with minimal medium change. In vitro generation of bona fide fully differentiated adipocytes was assessed by leptin secretion and ultrastructurally demonstrated through semithin to ultrathin sectioning and lipid staining with osmium tetroxide. Furthermore, presence of BM-MSCs or ASCs within the subcutaneous layer contributed to the epidermal differentiation program, with more proliferating basal cells depositing basal membrane proteins and differentiating into mature keratinocytes that were able to generate a pluristratified epithelium. In conclusion, we engineered a fully differentiated human skin trilayer that could present multiple applications such as use for in vitro drug absorption tests and regenerative therapies.
Collapse
|
45
|
Stem cells, mature adipocytes, and extracellular scaffold: what does each contribute to fat graft survival? Aesthetic Plast Surg 2011; 35:1061-72. [PMID: 21590499 DOI: 10.1007/s00266-011-9734-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 04/06/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Soft tissue engineering offers new perspectives for improving fat graft survival, for which the appropriate association of cells and scaffold seems essential. This study aimed to analyze the survival of free-cell grafts compared with adipose-derived stem cells (ASCs) seeded on collagen scaffolds. METHODS Adipose tissue from a single volunteer was used for the following preparations: purified adipose tissue, isolated mature adipocytes (free-cell graft), cultured ASCs without scaffold (free-cell graft), collagen scaffold only, cultured ASCs in collagen scaffold without and with bioactive factors, and freshly-isolated ASCs in collagen scaffold. These were grafted on 18 nude mice for 2 months, after which specimens were evaluated grossly and histologically using hematoxylin-phloxine-safran (HPS), Oil-Red-O, and antivimentin labeling. Specimens and animals were weighed before implantation and after explantation, and weight values were statistically analyzed. RESULTS Free-cell grafts (mature adipocytes and free ASCs) showed complete resorption in 50 and 60% of the animals (remaining weight fraction was 22.5 and 5.3%, respectively). The survival of purified adipose tissue was 81.8% (statistically greater compared with free-cell grafts; p < 0.05). In the ASCs-scaffold association, the remaining weight fractions (87.3-70.4%) were statistically greater than in free-cell grafts (5.3-22.5%; p < 0.05), but the difference between ASC-scaffolds and fat grafts was not statistically significant. These results were confirmed by clinical and histologic observations. CONCLUSION Three-dimensional collagen scaffolds seem to improve survival of ASCs compared with free-cell grafts (adipocytes and free ASCs).
Collapse
|
46
|
Mitsak AG, Dunn AM, Hollister SJ. Mechanical characterization and non-linear elastic modeling of poly(glycerol sebacate) for soft tissue engineering. J Mech Behav Biomed Mater 2011; 11:3-15. [PMID: 22658150 DOI: 10.1016/j.jmbbm.2011.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 01/01/2023]
Abstract
Scaffold tissue engineering strategies for repairing and replacing soft tissue aim to improve reconstructive and corrective surgical techniques whose limitations include suboptimal mechanical properties, fibrous capsule formation and volume loss due to graft resorption. An effective tissue engineering strategy requires a scaffolding material with low elastic modulus that behaves similarly to soft tissue, which has been characterized as a nonlinear elastic material. The material must also have the ability to be manufactured into specifically designed architectures. Poly(glycerol sebacate) (PGS) is a thermoset elastomer that meets these criteria. We hypothesize that the mechanical properties of PGS can be modulated through curing condition and architecture to produce materials with a range of stiffnesses. To evaluate this hypothesis, we manufactured PGS constructs cured under various conditions and having one of two architectures (solid or porous). Specimens were then tensile tested according to ASTM standards and the data were modeled using a nonlinear elastic Neo-Hookean model. Architecture and testing conditions, including elongation rate and wet versus dry conditions, affected the mechanical properties. Increasing curing time and temperature led to increased tangent modulus and decreased maximum strain for solid constructs. Porous constructs had lower nonlinear elastic properties, as did constructs of both architectures tested under simulated physiological conditions (wetted at 37 °C). Both solid and porous PGS specimens could be modeled well with the Neo-Hookean model. Future studies include comparing PGS properties to other biological tissue types and designing and characterizing PGS scaffolds for regenerating these tissues.
Collapse
Affiliation(s)
- Anna G Mitsak
- Department of Biomedical Engineering, 1101 Beal Avenue, Ann Arbor, MI 48104, USA
| | | | | |
Collapse
|
47
|
Creation of bony microenvironment with CaP and cell-derived ECM to enhance human bone-marrow MSC behavior and delivery of BMP-2. Biomaterials 2011; 32:6119-30. [PMID: 21632105 DOI: 10.1016/j.biomaterials.2011.05.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 05/05/2011] [Indexed: 11/21/2022]
Abstract
Extracellular matrix (ECM) comprises a rich meshwork of proteins and proteoglycans, which not only contains biological cues for cell behavior, but is also a reservoir for binding growth factors and controlling their release. Here we aimed to create a suitable bony microenvironment with cell-derived ECM and biodegradable β-tricalcium phosphate (β-TCP). More specifically, we investigated whether the ECM produced by bone marrow-derived mesenchymal stem cells (hBMSC) on a β-TCP scaffold can bind bone morphogenetic protein-2 (BMP-2) and control its release in a sustained manner, and further examined the effect of ECM and the BMP-2 released from ECM on cell behaviors. The ECM was obtained through culturing the hBMSC on a β-TCP porous scaffold and performing decellularization and sterilization. SEM, XPS, FTIR, and immunofluorescent staining results indicated the presence of ECM on the β-TCP and the amount of ECM increased with the incubation time. BMP-2 was loaded onto the β-TCP with and without ECM by immersing the scaffolds in the BMP-2 solution. The loading and release kinetics of the BMP-2 on the β-TCP/ECM were significantly slower than those on the β-TCP. The β-TCP/ECM exhibited a sustained release profile of the BMP-2, which was also affected by the amount of ECM. This is probably because the β-TCP/ECM has different binding mechanisms with BMP-2. The β-TCP/ECM promoted cell proliferation. Furthermore, the BMP-2-loaded β-TCP/ECM stimulated reorganization of the actin cytoskeleton, increased expression of alkaline phosphatase and calcium deposition by the cells compared to those without BMP-2 loading and the β-TCP with BMP-2 loading.
Collapse
|
48
|
Choi JH, Gimble JM, Lee K, Marra KG, Rubin JP, Yoo JJ, Vunjak-Novakovic G, Kaplan DL. Adipose tissue engineering for soft tissue regeneration. TISSUE ENGINEERING PART B-REVIEWS 2011; 16:413-26. [PMID: 20166810 DOI: 10.1089/ten.teb.2009.0544] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Current treatment modalities for soft tissue defects caused by various pathologies and trauma include autologous grafting and commercially available fillers. However, these treatment methods present a number of challenges and limitations, such as donor-site morbidity and volume loss over time. As such, improved therapeutic modalities need to be developed. Tissue engineering techniques offer novel solutions to these problems through development of bioactive tissue constructs that can regenerate adipose tissue in both structure and function. Recently, a number of studies have been designed to explore various methods to engineer human adipose tissue. This review will focus on these developments in the area of adipose tissue engineering for soft tissue replacement. The physiology of adipose tissue and current surgical therapies used to replace lost tissue volume, specifically in breast tissue, are introduced, and current biomaterials, cell sources, and tissue culture strategies are discussed. We discuss future areas of study in adipose tissue engineering.
Collapse
Affiliation(s)
- Jennifer H Choi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Higuchi A, Ling QD, Ko YA, Chang Y, Umezawa A. Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. Chem Rev 2011; 111:3021-35. [PMID: 21344932 DOI: 10.1021/cr1003612] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, 32001 Taiwan.
| | | | | | | | | |
Collapse
|
50
|
Choi JH, Bellas E, Vunjak-Novakovic G, Kaplan DL. Adipogenic differentiation of human adipose-derived stem cells on 3D silk scaffolds. Methods Mol Biol 2011; 702:319-30. [PMID: 21082412 DOI: 10.1007/978-1-61737-960-4_23] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Current treatment modalities for soft tissue defects due to various pathologies and trauma include autologous grafting and the use of commercially available fillers. However, these treatment methods are associated with a number of limitations, such as donor site morbidity and volume loss over time. As such, improved therapeutic options are needed. Tissue engineering techniques offer novel solutions to these problems through development of bioactive tissue constructs that can regenerate adipose tissue with an appropriate structure and function. The recent advances in the derivation and characterization of hASCs have led to numerous studies of soft tissue reconstruction. In this chapter, we discuss methods in which our laboratory has used hASCs and silk scaffolds for adipose tissue engineering. The use of naturally occurring and clinically acceptable materials such as silk protein for tissue-engineering applications poses advantages with respect to biocompatibility and mechanical and biological properties.
Collapse
|